Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
2.
In Vitr Mol Toxicol ; 14(2): 121-7, 2001.
Article in English | MEDLINE | ID: mdl-11690565

ABSTRACT

Bemitradine is a compound that was intended for use as a diuretic antihypertensive drug. In the preclinical safety assays, it was found to be nongenotoxic in five in vitro assays (Ames, mouse lymphoma, CHO/HGPRT, CHO chromosome aberration, and UDS) and in one in vivo assay (mouse bone marrow micronucleus). In a subsequent long-term bioassay using Sprague-Dawley rats, this compound was found to be a rodent carcinogen at multiple sites. Because of the carcinogenicity, further development of this compound as a drug was halted. Because the Syrian hamster embryo (SHE) cell transformation assay at pH 6.7 has been demonstrated to be a good predictor of carcinogenic activity in animals, this nongenotoxic compound was used to determine if this in vitro assay system could be utilized to predict the potential for the carcinogenicity in rodents. The SHE cell transformation assay was validated initially for use in this laboratory using genotoxic and nongenotoxic carcinogens including benzo(a)pyrene, 20-methylcholanthrene, 2-acetylaminofluorene, methapyrilene, and phenobarbital. Each of these chemicals induced a statistically significant increase in morphological transformation frequency. Bemitradine was initially tested in a range-finding cytotoxicity assay at 10-250 microg/mL for treatment periods of 7 days. Doses used in the 7-day treatment transformation assay were 1.25, 2.5, 5.0, 7.5, and 10.0 microg/mL. Statistically significant increases in morphological transformation frequencies were observed at 1.25, 2.5, and 7.5 microg/mL, indicating a positive response. The experiment was repeated with similar results confirming the previous conclusion. These data provide additional evidence that the pH 6.7 SHE cell transformation assay may be a valuable in vitro tool to detect nongenotoxic rodent carcinogens.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Pyrimidines/administration & dosage , Triazoles/administration & dosage , Animals , Carcinogenicity Tests/methods , Cricetinae , Embryo, Mammalian , Hydrogen-Ion Concentration , Mesocricetus
3.
Toxicol Pathol ; 29 Suppl: 138-46, 2001.
Article in English | MEDLINE | ID: mdl-11695550

ABSTRACT

The Syrian hamster embryo (SHE) cell-transformation assay represents a short-term in vitro assay capable of predicting rodent carcinogenicity of chemicals with a high degree of concordance (LeBoeuf et al [1996]. Mutat Res 356: 85-127). The SHE assay models the earliest identifiable stage in carcinogenicity, morphological cell transformation. In contrast to other short-term in vitro assays, both genotoxic and epigenetic carcinogens are detected. The SHE assay, originally developed by Berwald and Sachs (J Natl Cancer Inst 35: 641-661) and modified as described by LeBoeuf and Kerckaert (Carcinogenesis 7: 1431-1440), was included in the International Life Sciences Institute, Health and Environmental Sciences Institute (ILSI/HESI). Alternative Carcinogenicity Testing (ACT) collaboration to provide additional information on the use of short-term in vitro tests in predicting carcinogenic potential. A total of 19 ILSI compounds have been tested in the SHE assay: 15 were tested for this project, whereas clofibrate, methapyrilene, reserpine, and Di(2-ethylhexyl)phalate (DEHP) were tested previously. Of the 3 noncarcinogenic compounds tested, 2 were negative in the SHE assay, whereas ampicillin was tested positive. The remaining 16 compounds tested were either known rodent carcinogens and/or human carcinogens. From this group, 15 tested positive in the SHE assay whereas phenacetin, a genotoxic carcinogen, was tested negative. Therefore, overall concordance between the SHE assay and rodent bioassay was 89% (17/19), whereas concordance with known or predicted human carcinogens was 37% (7/19). Based on these data, it is concluded that the SHE cell-transformation assay has utility for predicting the results of the rodent carcinogenesis bioassay but lacks the selectivity to distinguish between rodent and human carcinogens.


Subject(s)
Carcinogenicity Tests/methods , Carcinogens/toxicity , Cell Transformation, Neoplastic/chemically induced , Embryo, Mammalian/drug effects , Mutagens/toxicity , Animal Testing Alternatives , Animals , Cricetinae , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , In Vitro Techniques , Mesocricetus , Mice , Predictive Value of Tests , Rats , Reproducibility of Results
4.
J Histochem Cytochem ; 49(7): 927-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11410620

ABSTRACT

In this study we evaluated whether storing non-deparaffinized sections can affect the detection of specific mRNAs by radioactive in situ hybridization (ISH). Using a standard ISH protocol, we hybridized serial sections of paraffin blocks stored for different periods of time with (33)P-labeled riboprobes specific for rat Type III collagen and matrix metalloproteinase-2 (MMP-2). Signal intensities were evaluated using a phosphorimager and by blinded microscopic examination. For slides hybridized with the Type III collagen riboprobe, signal intensities measured with the phosphorimager or evaluated by microscopic examination were negatively correlated with the storage period of the sections. For slides hybridized with the MMP-2 riboprobe, differences in signal intensity could be detected, albeit inconsistently, with the phosphorimager, although microscopic examination consistently indicated stronger signals in freshly sectioned slides compared to slides stored for 2 weeks or more. We concluded that it was preferable to use recently prepared sections for trying to locate mRNAs in paraffin-embedded tissues by ISH. In addition, our results suggest that quantifying signal intensity using a phosphorimager is feasible for abundant mRNAs or when large differences in expression are anticipated.(J Histochem Cytochem 49:927-928, 2001)


Subject(s)
Paraffin Embedding , RNA, Messenger/analysis , Animals , Bleomycin , Collagen/analysis , Collagen/genetics , Collagen/metabolism , In Situ Hybridization/methods , Lung/metabolism , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Myocardial Infarction/metabolism , Phosphorus Radioisotopes , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism , Rats , Skin/injuries , Skin/metabolism , Time Factors
5.
Toxicol Pathol ; 27(1): 104-6, 1999.
Article in English | MEDLINE | ID: mdl-10367682

ABSTRACT

Significant change is occurring in the drug discovery paradigm; many companies are utilizing dedicated groups from the toxicology/ pathology disciplines to support early stage activities. The goal is to improve the efficiency of the discovery process for selecting a successful clinical candidate. Toxicity can be predicted by leveraging molecular techniques via rapid high-throughput, low-resource in vitro and in vivo test systems. Several important activities help create a platform to support rapid development of a new molecular entity. The proceedings of this symposium provide excellent examples of these applied concepts in pharmaceutical research and development. Leading biopharmaceutical companies recognize that a competitive advantage can be maintained via rapid characterization of animal models, the cellular identification of therapeutic targets, and improved sensitivity of efficacy assessment. The participation of the molecular pathologist in this quest is evolving rapidly, as evidenced by the growing number of pathologists that interact with drug discovery organizations.


Subject(s)
Biopharmaceutics , Drug Design , Pathology/methods , Toxicology , Animals , Pharmacology/methods
6.
Toxicol Pathol ; 27(1): 107-10, 1999.
Article in English | MEDLINE | ID: mdl-10367683

ABSTRACT

The remarkable technologic and methodologic advances spurred on by the Human Genome Project are being applied throughout the life sciences. In the field of toxicology, high-resolution assays now make it possible to discover virtually all the differences in gene expression brought on by exposure to a particular xenobiotic. There are 2 principal approaches used to build a catalog of changes in gene expression: hybridization microarrays and gel-based methods, such as differential display and AFLP-based mRNA finger-printing. The power of such approaches is exemplified by the identification of more than 300 genes that differ in expression level by at least 2-fold in response to the nongenotoxic rodent liver carcinogen phenobarbital.


Subject(s)
Genome, Human , Toxicology , Animals , Gene Expression Regulation/drug effects , Genetic Techniques , Humans
7.
Toxicol Pathol ; 26(5): 612-20, 1998.
Article in English | MEDLINE | ID: mdl-9789947

ABSTRACT

Cyclooxygenase (COX) exists in 2 related but unique isoforms: one is constitutive (COX-1) and functions in normal cell physiology, and the other is inducible (COX-2) and is expressed in response to inflammatory stimuli. Nonsteroidal antiinflammatory drugs (NSAIDs) cause renal toxicity following inhibition of renal cyclooxygenases. Humans and animals exhibit differences in susceptibility to NSAID-related renal toxicity, which may be associated with differences in expression of 1 or both isoforms of COX in the kidney. In this study, we evaluated COX-1 and COX-2 expression in the kidneys of mixed-breed dogs, Sprague-Dawley rats, cynomolgus monkeys, and humans. In addition, the effect of volume depletion on renal COX expression was investigated in rats, dogs, and monkeys. COX expression was evaluated using 1 or more of the following procedures: reverse transcriptase polymerase chain reaction, in situ hybridization, and immunohistochemistry. We demonstrated that both COX isoforms are expressed in the kidneys of all species examined, with differences in the localization and level of basal expression. COX-1 is expressed at high levels in the collecting ducts and renal vasculature of all species and in a small number of papillary interstitial cells in rats, monkeys, and humans. Basal levels of COX-2 are present in the maculae densa, thick ascending limbs, and papillary interstitial cells in rats and dogs and in glomerular podocytes and small blood vessels in monkeys and humans. COX-2 expression is markedly increased in volume-depleted rats and dogs but not monkeys. These results indicate that significant interspecies differences exist in the presence and distribution of COX isoforms, which may help explain the difference in species susceptibility to NSAID-related renal toxicity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Isoenzymes/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Kidney/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Adult , Animals , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/toxicity , Dogs , Humans , Immunohistochemistry , In Situ Hybridization , Macaca fascicularis , Membrane Proteins , Middle Aged , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Species Specificity
8.
Toxicol Pathol ; 26(4): 492-500, 1998.
Article in English | MEDLINE | ID: mdl-9715508

ABSTRACT

The tumorigenic potential of phenobarbital was examined in a 26-wk carcinogenesis bioassay using p53 heterozygous mice and wild-type controls. Fifteen mice/sex/genotype were exposed to either 500 or 1,000 ppm phenobarbital in the diet. Dietary administration of 3,750 ppm p-cresidine, a transspecies mutagenic carcinogen, to both heterozygous and wild-type mice served as a positive control. Phenobarbital treatment caused increases in liver:body weight ratios and histologic evidence of centrilobular hepatocellular hypertrophy. No tumors were observed in any phenobarbital-treated mice. Mice given p-cresidine exhibited a moderate reduction in body weight gain over the course of the study. Heterozygous mice treated with p-cresidine exhibited a high incidence of urinary bladder tumors. Similar tumors were also present in a small number of p-cresidine-treated wild-type mice. Our results demonstrate the lack of a hepatic tumor response to phenobarbital, a compound that is a potent and potent and prototypic hepatic microsomal enzyme inducer, a nongenotoxic rodent carcinogen, and a human noncarcinogen. This finding supports the continued utility of this model as an alternative to the mouse bioassay for human carcinogenic safety assessment of potentially genotoxic carcinogenes because it did not produce a false-positive response to this potent nongenotoxic agent.


Subject(s)
Carcinogens/toxicity , Genes, p53/genetics , Hypnotics and Sedatives/toxicity , Liver Neoplasms, Experimental/chemically induced , Phenobarbital/toxicity , Aniline Compounds/toxicity , Animals , Body Weight/drug effects , Female , Heterozygote , Liver/pathology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Survival Analysis
9.
Cell Growth Differ ; 8(7): 779-88, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9218872

ABSTRACT

A variety of agents, such as caffeine, have been shown to abrogate the DNA damage-dependent G2 checkpoint and enhance cytotoxicity. However, these agents are too toxic for clinical use. We have reported that the potent protein kinase inhibitor 7-hydroxystaurosporine (UCN-01) at nontoxic doses abrogates the G2 arrest caused by the DNA-damaging agent cisplatin. Here, using Chinese hamster ovary cells, we show that cisplatin causes predominantly an S-phase arrest; UCN-01 abrogates this S-phase arrest, causing progression of cells to G2 and, subsequently, apoptotic cell death. In searching for an explanation for this accelerate DNA synthesis, we discovered that UCN-01 caused translocation of proliferating cell nuclear antigen (PCNA) to the detergent-insoluble, DNA-bound fraction. PCNA acts as a sliding clamp for DNA polymerase delta. Sequestering of PCNA by p21waf1/cip1 is required for p53-dependent G1 arrest in damaged cells. However, the S-phase arrest occurs independently of p53 and p21waf1/cip1. Our results suggest that PCNA is also a component of this S-phase checkpoint, despite the fact that CHO cells are defective for p53, and no increase in p21waf1/cip1 was observed. The mechanism by which PCNA is sequestered in the absence of p21waf1/cip1 and the mechanism by which UCN-01 disrupts this sequestration remain to be elucidated.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinase Inhibitors , S Phase/drug effects , Animals , Aphidicolin/pharmacology , CDC2 Protein Kinase/metabolism , CHO Cells , Cell Fractionation , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , DNA/biosynthesis , DNA Fragmentation/drug effects , Enzyme Inhibitors/pharmacology , G2 Phase/physiology , Phosphorylation , Staurosporine/analogs & derivatives , Tumor Suppressor Protein p53/physiology
10.
Clin Cancer Res ; 2(5): 791-7, 1996 May.
Article in English | MEDLINE | ID: mdl-9816232

ABSTRACT

DNA-damaging agents arrest cell cycle progression at either G1 or G2. A variety of agents such as caffeine have been shown to abrogate the DNA damage-dependent G2 checkpoint and enhance cytotoxicity. Unfortunately, this strategy has not enhanced therapeutic activity because adequate concentrations of these modulators are not tolerated in vivo. Here, using Chinese hamster ovary cell lines, we show that the potent protein kinase inhibitor 7-hydroxy-staurosporine (UCN-01) abrogates the G2 arrest induced by the DNA-damaging agent cisplatin. UCN-01 not only was effective at inducing mitosis when added to G2-arrested cells but also prevented cells from arresting in G2 when added to S-phase cells. Furthermore, UCN-01 did not cause premature mitosis of S-phase cells; rather, the cells progressed to G2 before undergoing mitosis. These effects were observed at noncytotoxic concentrations of UCN-01 that alone had no effect on cell cycle passage. Furthermore, the same concentrations of UCN-01 that resulted in abrogation of the cisplatin-induced G2 arrest also enhanced cisplatin-induced cytotoxicity, as determined by a colony formation assay. UCN-01 enhanced cisplatin cytotoxicity up to 60-fold and reduced by 3-fold the concentration of cisplatin required to kill 90% of the cells. The concentrations of UCN-01 required for this enhancement have been shown to be well tolerated in animal models, suggesting that this combination may represent an effective strategy for enhancing cisplatin-based chemotherapeutic regimens.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , G2 Phase/drug effects , Protein Kinase C/antagonists & inhibitors , Animals , CHO Cells , Cell Survival/drug effects , Cricetinae , Drug Synergism , Staurosporine/analogs & derivatives
11.
Int J Radiat Biol ; 68(5): 553-62, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7490505

ABSTRACT

A recently developed, combined alkaline unwinding/Southern blotting assay was utilized to examine DNA damage and repair induced by ionizing radiation within specific large-scale genomic regions. Following treatment of MCF-7 breast tumour cells with 2-10-Gy gamma-rays, strand breakage and rejoining were measured in bulk DNA, in the centromeric alpha-satellite region of chromosome 17, and in the chromatin regions containing the unexpressed beta-globin gene and the expressed c-myc oncogene, which is known to be important for growth in the MCF-7 cell line. Damage in both the c-myc and beta-globin regions was markedly greater than in either alpha-satellite or bulk DNA. However, the kinetics of strand break repair were approximately the same in c-myc as in alpha-satellite or bulk DNA. Surprisingly, the radiomimetic antibiotic bleomycin, which also induces free-radical-mediated strand breakage, showed considerably less heterogeneity of DNA damage among the genomic regions examined than did radiation. The results suggest that actively transcribed genes, as well as at least some inactive genes, are surrounded by large-scale domains of radiosensitive chromatin. With no apparent enhancement of rejoining, the increased incidence of strand breaks in these regions persists until rejoining is essentially complete. Changes in the integrity of specific chromatin regions may be an important aspect of DNA damage-induced cell death.


Subject(s)
DNA Damage , DNA Repair , DNA/radiation effects , Bleomycin/toxicity , Blotting, Southern , DNA/drug effects , Female , Humans , Tumor Cells, Cultured
12.
Biochem Pharmacol ; 50(8): 1317-9, 1995 Oct 12.
Article in English | MEDLINE | ID: mdl-7488252

ABSTRACT

Previous studies with MCF-7 cells demonstrated that several agents induce greater strand breakage in active genes than in nontranscribed centromeric regions. To better assess the effects of gene activity and inactivity, an allele-specific DNA strand break assay was developed, which allowed direct comparison of damage at a specific genetic locus on the active and inactive X chromosomes. The ZP lymphoblastoid cell line is heterozygous at the glucose-6-phosphate dehydrogenase (G6PD) locus, and the unexpressed (A) allele on the inactive X chromosome contains a FokI restriction site that is lacking in the expressed (B) allele on the active X. ZP cells were treated with camptothecin or amsacrine, and subjected to alkaline-induced DNA unwinding. Following detergent lysis and digestion of single-stranded DNA with S1 nuclease, the remaining double-stranded DNA was isolated and subjected to polymerase chain reaction (PCR) with primers that flank the polymorphic FokI site, with [alpha-32P]dCTP being added in the last PCR cycle. The resulting labeled PCR product was cleaved with FokI to assess the A/B allele ratio in the double-stranded DNA fraction. Treatment with camptothecin and amsacrine increased the apparent A/B ratio by factors of 2-3 and 1.5-2 respectively, indicating that the active B allele is preferentially damaged by these agents.


Subject(s)
Alleles , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Lymphocytes/drug effects , X Chromosome/drug effects , Amsacrine/pharmacology , Base Sequence , Cell Line, Transformed , DNA/isolation & purification , DNA Damage , Genetic Techniques , Glucosephosphate Dehydrogenase/genetics , Humans , Hydrogen-Ion Concentration , Lymphocytes/ultrastructure , Molecular Sequence Data
13.
Cell Growth Differ ; 5(9): 983-93, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7819136

ABSTRACT

Many anticancer drugs arrest cells in G2 of the cell cycle and subsequently induce cell death by apoptosis. The current experiments establish a detailed sequence of events that occur in Chinese hamster ovary CHO/UV41 cells following incubation with cisplatin. Synchronized CHO/UV41 cells were damaged with cisplatin in early S. The cells progressed at a normal rate through S but arrested in G2. The arrested cells exhibited normal levels of the mitosis-promoting kinase p34cdc2 in its fully phosphorylated, inactive form. After a protracted arrest, the cells dephosphorylated p34cdc2 and underwent an aberrant mitosis and cytokinesis in which the chromosomes segregated unequally due to the formation of multipolar mitotic spindles. These cells subsequently lost contact with the extracellular matrix, and only then digested their DNA in a manner characteristic of apoptosis. This sequence of events could be dramatically accelerated by the addition of caffeine to G2-arrested cells, which induced dephosphorylation of p34cdc2 and passage through an aberrant mitosis. It has previously been suggested that protein synthesis is required for both caffeine-induced premature mitosis and apoptosis. However, when added in G2, cycloheximide could inhibit neither the caffeine-induced mitosis nor apoptosis. Inhibition was only seen if cycloheximide was added during S before complete synthesis of the proteins required for mitosis. These results demonstrate that, in this model, the proteins thought to be involved in apoptosis are those required for normal cell cycle progression. It is hypothesized that the DNA digestion results from loss of signal transduction originating from the extracellular matrix but that earlier events leading to loss of cell adhesion are critical for the induction of apoptosis.


Subject(s)
Apoptosis/drug effects , CHO Cells/drug effects , Cell Cycle/drug effects , Cisplatin/toxicity , Animals , CDC2 Protein Kinase/metabolism , Caffeine/pharmacology , Cell Division/drug effects , Cricetinae , DNA/drug effects , Mitosis/drug effects , Phosphorylation , Protein Biosynthesis
14.
Biochem Pharmacol ; 47(2): 317-29, 1994 Jan 20.
Article in English | MEDLINE | ID: mdl-8304976

ABSTRACT

In the MCF-7 human breast tumor cell line, the aminoacridine, m-AMSA, induces protein-associated DNA strand breaks consistent with inhibition of topoisomerase II. However, neither single-strand nor double-strand breaks in DNA, determined using conventional assays, show a consistent relationship with m-AMSA-induced inhibition of growth. In contrast, when DNA strand breaks are determined by alkaline unwinding under the high salt conditions of the alkaline unwinding/Southern blotting (AU/SB) assay, developed by our laboratories, damage to DNA corresponds closely with growth inhibition. The AU/SB assay, which is capable of assessing breaks within large-scale domains (upwards of 1 megabase) surrounding genes of interest, was further utilized to explore the capacity of m-AMSA to induce damage within specific genomic regions that may regulate cell growth. Regions encompassing the transcriptionally active oncogenes, c-myc and c-fos, were found to be more susceptible to m-AMSA-induced strand breaks than the region encompassing the non-transcribed alpha-satellite DNA or the genome as a whole (bulk DNA). These findings demonstrate that m-AMSA may produce more pronounced damage within specific genomic regions than in bulk DNA, m-AMSA also preferentially altered expression of the c-myc oncogene; at an m-AMSA concentration where growth was inhibited by between 70 and 80%, steady-state c-myc mRNA levels declined to approximately 10-15% of control levels within 2-3 hr; furthermore, concentration-dependent reductions in c-myc expression appeared to coincide with growth inhibition. In addition, inhibition of [3H]thymidine incorporation after 2 hr directly paralleled inhibition of growth, suggesting an early effect at the level of DNA biosynthesis, possibly related to the down-regulation of c-myc expression. It is proposed that specific lesions, e.g., in regions surrounding the c-myc gene, as well as generalized lesions in DNA may lead to growth inhibition mediated by down-regulation of the expression of select growth regulatory genes, such as c-myc.


Subject(s)
Amsacrine/pharmacology , DNA Damage , DNA/drug effects , Cell Division/drug effects , DNA, Satellite/drug effects , DNA, Single-Stranded/drug effects , Down-Regulation , Gene Expression/drug effects , Genes, myc/drug effects , Humans , RNA, Messenger/analysis , Tumor Cells, Cultured/drug effects
15.
Cancer Res ; 53(15): 3547-54, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-8339261

ABSTRACT

In the MCF-7 breast tumor cell line, induction of bulk damage to DNA (measured either as total strand breaks or as double-strand breaks) fails to correspond with the antiproliferative activity of the demethylepipodo-phyllotoxin derivative, VM-26. In contrast, VM-26 produces an early (within 2-3 h) concentration-dependent reduction in c-myc expression (and of DNA synthesis) which parallels inhibition of cell growth, suggesting the possibility of effects of VM-26 at the level of genomic regions which regulate DNA replicative function. Although VM-26 also produces a reduction in c-myc expression in K562 human leukemic cells, these alterations fail to correspond with the concentration-dependent effects on cell growth in this cell line. Utilizing the newly developed alkaline unwinding/Southern blotting assay in the MCF-7 breast tumor cell line, it was determined that VM-26 induces damage within regions surrounding the c-myc gene and the beta-globin gene which exceeds that induced in both alpha-satellite DNA and in L1 repeat sequences; damage within c-myc and beta-globin also exceeds that observed throughout the genome as a whole. These findings indicate that certain genomic regions incur preferential damage in MCF-7 cells exposed to VM-26. It appears possible that damage within such genomic regions could lead to alterations in expression of select genes associated with regulation of cellular proliferation, resulting in reduced DNA synthesis, compromised cell growth, and, ultimately, cell death.


Subject(s)
Breast Neoplasms/genetics , DNA Damage , DNA, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Teniposide/pharmacology , Blotting, Southern , Breast Neoplasms/pathology , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Female , Genes, myc , Humans , RNA, Neoplasm/biosynthesis , Tumor Cells, Cultured
16.
Oncol Res ; 4(1): 7-15, 1992.
Article in English | MEDLINE | ID: mdl-1581618

ABSTRACT

A method is described which combines alkaline induced DNA unwinding and Southern blotting to measure DNA damage occurring in specific genomic regions. Damage induced by gamma-rays at levels as low as 2 Gy was measured in bulk DNA and in a one megabase region surrounding the actively transcribed oncogene, c-myc, as well as in the inactive alpha-satellite DNA of chromosome 17. Although the unwinding kinetics for bulk DNA were consistent with random strand breakage throughout the genome as a whole, measurements at specific loci indicated that the region encompassing c-myc was at least 2-fold more susceptible to damage than either the bulk of the genome or the alpha-satellite region. The results of this study indicate that the combined alkaline unwinding/Southern blotting assay is a sensitive method for the detection of DNA damage within specific chromatin regions, at biologically relevant doses.


Subject(s)
DNA Damage , DNA/radiation effects , Genes, myc/radiation effects , Blotting, Southern , DNA, Satellite , Gamma Rays , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Tumor Cells, Cultured
17.
Biochem Pharmacol ; 43(2): 331-42, 1992 Jan 22.
Article in English | MEDLINE | ID: mdl-1310853

ABSTRACT

A carcinogen-transformed rat hepatoma cell line (Reuber H-35) was utilized as a model system for investigation of the biochemical factors which may limit the effectiveness of chemotherapy in intrinsically resistant tumors such as hepatocellular carcinoma. Northern blotting demonstrated expression of mRNA coding for the P-170 membrane-glycoprotein associated with the multi-drug resistance phenotype, while Western blotting identified the P-170 glycoprotein in the hepatoma cell membrane. Consistent with these observations, tumor cell sensitivity to the vinca alkaloids, vincristine and vinblastine, to the anthracycline antibiotics, Adriamycin and daunorubicin, and to the demethylepipodophyllotoxin derivative, VM-26, was enhanced by continuous incubation in the presence of the calcium channel antagonist, verapamil. Verapamil produced a minimal change in cell sensitivity to the demethylepipodophyllotoxin derivative, VP-16, and to the aminoacridine, m-AMSA. Relatively high detoxification potential via the glutathione metabolic pathway was also observed in the hepatoma cell. The capacity of topoisomerase II in nuclear extracts from the hepatoma cell to mediate cleavable complex formation stimulated by VM-26, VP-16 and m-AMSA appeared to be at least comparable to, if not greater than that from drug-sensitive HL-60 cells, suggesting that drug resistance may not occur at the level of this enzyme. Consistent with findings in a number of tumor cell lines resistant to antineoplastic drugs, the antiproliferative activity of the topoisomerase II inhibitors VM-26, VP-16 and m-AMSA appeared to be dissociable from the induction of DNA strand breaks, suggesting that such lesions in DNA may fail to fully account for the antiproliferative activity of these agents in the hepatoma cell.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance/genetics , Liver Neoplasms, Experimental/genetics , Animals , Binding Sites/drug effects , Blotting, Northern , Blotting, Western , Cell Division/drug effects , Cell Fractionation , Cell Line/drug effects , DNA Damage , Glutathione/metabolism , Liver Neoplasms, Experimental/enzymology , Membrane Glycoproteins/metabolism , Phenotype , Rats , Topoisomerase II Inhibitors , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL